Although the central nervous system has been implicated in glucocorticoid-induced gain of fat mass, the underlying mechanisms are poorly understood. The aim of this study was to investigate the possible involvement of hypothalamic serum- and glucocorticoid-regulated kinase 1 (SGK1) in glucocorticoid-increased adiposity. It is well known that SGK1 expression is induced by acute glucocorticoid treatment, but it is interesting that we found its expression to be decreased in the arcuate nucleus of the hypothalamus, including proopiomelanocortin (POMC) neurons, following chronic dexamethasone (Dex) treatment. To study the role of SGK1 in POMC neurons, we produced mice that developed or experienced adult-onset SGK1 deletion in POMC neurons (PSKO). As observed in Dex-treated mice, PSKO mice exhibited increased adiposity and decreased energy expenditure. Mice overexpressing constitutively active SGK1 in POMC neurons consistently had the opposite phenotype and did not experience Dex-increased adiposity. Finally, Dex decreased hypothalamic α-melanocyte-stimulating hormone (α-MSH) content and its precursor Pomc expression via SGK1/FOXO3 signaling, and intracerebroventricular injection of α-MSH or adenovirus-mediated FOXO3 knockdown in the arcuate nucleus largely reversed the metabolic alterations in PSKO mice. These results demonstrate that POMC SGK1/FOXO3 signaling mediates glucocorticoid-increased adiposity, providing new insights into the mechanistic link between glucocorticoids and fat accumulation and important hints for possible treatment targets for obesity.

Despite the overwhelming beneficial anti-inflammatory effects of glucocorticoid, chronic glucocorticoid treatment has been shown to cause numerous adverse metabolic outcomes, including fat mass gain (1). Recent studies have elucidated several peripheral mechanisms underlying glucocorticoid-induced increase in fat mass. For example, glucocorticoid induces adipocyte differentiation (13), alters lipid metabolism in adipose tissue (13), and inhibits browning of white adipose tissue (WAT) or thermogenesis of brown adipose tissue (BAT) (4,5). In fact, body fat mass is largely controlled by the central nervous system (CNS) (68). Specific populations of neurons in the arcuate nucleus (ARC) of the hypothalamus also play fundamental roles in the regulation of energy balance and lipid metabolism (68). In particular, neurons coexpressing the orexigenic neuropeptides agouti-related protein and neuropeptide Y, and neurons coexpressing the anorexigenic proopiomelanocortin (POMC) precursor and the cocaine- and amphetamine-related transcript, are extensively involved in the regulation of appetite, body weight, and metabolism (68). POMC is a protein expressed and secreted from POMC neurons and cleaved by prohormone convertases to produce α-melanocyte-stimulating hormone (α-MSH) (8). α-MSH binds to the melanocortin 4 receptor and functions as a key hub linking the CNS to peripheral organs through the sympathetic nervous system (SNS), whereas dysfunction of this signaling axis leads to obesity in mice and humans (9,10). Activation of the SNS promotes the release of norepinephrine (NE) that binds to β-adrenergic receptor 3 and stimulates WAT lipolysis and BAT thermogenesis (1114). Although previous studies have shown that glucocorticoid regulates food intake and energy expenditure (15,16), the central signals mediating the effect of glucocorticoid are poorly understood.

Serum- and glucocorticoid-regulated kinase 1 (SGK1) belongs to the family of serine/threonine kinases, and its coding region was originally isolated from rat mammary tumor cells (17). SGK1 is ubiquitously expressed in various tissues, including hypothalamus (17), and functions via activation of the glucocorticoid receptor (GR), retinoid X receptor, peroxisome proliferator–activated receptor γ, and nuclear factor κB (17). SGK1 is involved in the regulation of many processes, including hypertension, epithelial sodium channel activity, and insulin sensitivity (1719). SGK1 also mediates many important functions of glucocorticoids, including insulin secretion and hippocampal neurogenesis (20,21). Although extensive studies have been carried out, a role for hypothalamic SGK1 in the regulation of energy homeostasis is unknown. Furthermore, SGK1 is well known as an early-response gene that can be induced by acute glucocorticoid treatment in various cells and animal models (2022); however, the effect of chronic glucocorticoid treatment on SGK1 expression remains largely unknown. In fact, the expression of SGK1 in the context of glucocorticoid-induced metabolic effects could be very important, as studies show that sometimes SGK1 may have effects opposing those of glucocorticoid (23).

Despite the aforementioned unknown facts, we can speculate that SGK1, as a downstream target of glucocorticoid (17) expressed in the hypothalamus (17), may contribute to the central action of glucocorticoid. Therefore, the aim of this study was to test this hypothesis first by determining the expression of SGK1 in the hypothalamus and then by investigating its possible contribution to glucocorticoid-increased adiposity.

By creating mice that develop or experience adult-onset knockout of SGK1, or overexpression of SGK1 in POMC neurons, we demonstrate a crucial role for SGK1 expressed in POMC neurons in glucocorticoid-increased adiposity and provide a novel mechanistic link between glucocorticoid treatment and body fat mass gain.

Mice and Diets

POMC-Cre (24) and tamoxifen-inducible POMC-cre (POMC-cre:ERT2) (24) mice were kindly provided by Joel K. Elmquist and Tiemin Liu from University of Texas Southwestern Medical Center (Dallas, TX); floxed SGK1 allele (SGK1loxp/loxp) mice (18) were provided by Dr. Geza Fejes-Toth and Dr. Aniko Naray-Fejes-Toth (Geisel School of Medicine, Dartmouth College, Hanover, NH). To generate POMC neuron–specific SGK1 knockout (PSKO) mice, POMC-Cre mice were crossed with SGK1loxp/loxp mice. To generate inducible POMC-specific SGK1 knockout mice, POMC-cre:ERT2 mice were crossed with SGK1loxp/loxp mice. Tamoxifen (0.15 g/kg; Sigma-Aldrich, St. Louis, MO) suspended in corn oil (Sigma-Aldrich) was intraperitoneally injected into 8-week-old male SGK1loxp/loxp or SGK1loxp/loxp × POMC-cre:ERT2 littermate mice for five consecutive days to generate mice with adult-onset SGK1 deletion in POMC neurons (PSKO-ER). Dexamethasone (Dex) treatment was administered to male wild-type (WT), control, and PSKO mice, and to male POMC-Cre mice injected with adeno-associated virus (AAV) expressing constitutively active mutant rat SGK1 (S422D) (AAV-CA SGK1)/AAV-null in ARC (control/POMC neuron–specific SGK1 overexpression [PSOE] mice) by intraperitoneal injection of PBS or 5 mg/kg Dex every other day for 6 weeks or for 2 h (1,25). All the mice were on a C57BL/6J background. Mice were maintained under a 12-h light/12-h dark cycle (lights on at 0700 h/lights off at 1900 h) at 25°C, with free access to water and standard chow diet. In vivo studies were conducted in accordance with the guidelines of the Institutional Animal Care and Use Committee of Shanghai Institute for Nutritional Sciences, Chinese Academy of Sciences.

Intracerebroventricular Cannulation and ARC Administration Experiments

Intracerebroventricular (ICV) cannulation experiments were conducted as previously described (26). After surgery, the mice were given 5 days to recover and were then infused with 2 μL α-MSH peptide (Abcam, Cambridge, U.K.) at a concentration of 1 nmol/μL or 2 μL artificial cerebrospinal fluid (Tocris, Bristol, U.K.), and experiments were conducted 24 h later. ARC administration experiments were conducted as previously described (6). Mice were anesthetized and received bilateral stereotaxic injections of adenovirus expressing FOXO3-specific short hairpin RNA against FOXO3 (Ad-shFOXO3) or scrambled control adenovirus (Ad-scrambled), AAV-CA SGK1, or control AAV-null into the ARC (1.5 mm posterior to the bregma, ±0.2 mm lateral to the midline, and 6 mm below the surface of the skull). The AAV-CA SGK1 expression plasmid was constructed in pAAV-Ef1a-DIO-mCherry-2A plasmid (Addgene, Cambridge, MA), and SGK1 started to express only in the presence of CRE recombinase.

Metabolic Parameter Measurements

The body composition of mice was measured with a nuclear magnetic resonance system (Bruker, Rheinstetten, Germany). Indirect calorimetry was performed in a comprehensive laboratory animal-monitoring system (Columbus Instruments, Columbus, OH), as previously described (27). Rectal temperature of mice was measured at 1400 and 1700 h with a rectal probe attached to a digital thermometer (Physitemp Instruments Inc., Clifton, NJ). Food intake was measured as reported previously (6).

POMC Neuron Identification, Count, and Area

AI9 (tdTomato) reporter mice (The Jackson Laboratory) were mated with, or AAV-CA SGK1 and AAV-null expressed mCherry red fluorescent protein were injected into the ARC of, POMC-Cre mice to reflect POMC neurons, demonstrated by co-localization with POMC antibodies. The distribution and number of POMC neurons were determined as described previously (6). Average somatic area was analyzed in >500 POMC neurons (n = 4 mice/genotype). The area occupied by POMC neurons was manually scored using ImageJ software.

Hypothalamic α-MSH Protein Content

Hypothalamus was prepared as previously described (6), and α-MSH was quantified with an ELISA kit (Phoenix Pharmaceuticals Inc., Burlingame, CA), according to the manufacturer’s instructions.

Hypothalamic Nuclear and Cytoplasmic Fractions

Hypothalamic nuclear and cytoplasmic fractions were isolated as previously described (28).

Immunofluorescence Staining

Immunofluorescence staining was performed, as previously described (29), with anti-SGK1 and the anti-p-N-myc downstream-regulated gene 1 (p-NDRG1) (Abcam), anti-POMC (Phoenix Pharmaceuticals Inc.), anti-FOXO3 (Cell Signaling Technology, Danvers, MA), anti-p-SGK1 and anti-GR (Santa Cruz Biotechnology, Santa Cruz, CA), and anti-α-MSH (Merck Millipore, Frankfurter, Germany). Immunofluorescence staining of p-FOXO3 was performed using the Tyramide Signal Amplification Cyanine 3 system (Perkin-Elmer, Boston, MA), and anti-p-FOXO3 primary antibody (Cell Signaling Technology) was coincubated with anti-mCherry (Abbkine, Inc., Wuhan, China).

RNA Isolation and Relative Quantitative RT-PCR

RNA was isolated and RT-PCR performed as previously described (27). The sequences of primers used in this study are available upon request.

Western Blotting Analysis

Western blotting was analyzed, as previously described (27), with the following primary antibodies: anti-p-FOXO3, anti-FOXO3, anti–lamin B1, and anti-p-GR (Cell Signaling Technology); anti-SGK1 and anti-GR (Abcam); anti–uncoupling protein 1 (UCP1) and anti-p-SGK1 (Santa Cruz Biotechnology); and anti-α-tubulin and anti-β-actin (Sigma-Aldrich).

Isolation and Treatment of Primary Hypothalamic Neurons

Primary cultures of hypothalamic neurons were prepared as previously described (27). On day 7, primary cultured hypothalamic neurons were infected with adenovirus expressing SGK1-specific short hairpin RNA (108 plaque-forming units/cells on 60 cm2) or Ad-scrambled, constructed as described previously (19). Primary hypothalamic neurons were transfected with small interfering RNA for FOXO3 using X-tremeGENE siRNA Transfection Reagent (Roche Diagnostics, Mannheim, Germany). Constitutively active mutant rat SGK1 (S422D) was subcloned into a PCMV-MYC plasmid and transfected into primary cultured hypothalamic neurons using Lipofectamine 2000 (Life Technologies).

Statistical Analysis

All values are presented as the mean ± SEM. Differences between groups were analyzed by either the Student t test or one-way ANOVA followed by the Student-Newman-Keuls test. Differences for which P was <0.05 were considered statistically significant.

Chronic Dex Treatment Decreases SGK1 Expression in Hypothalamic POMC Neurons

To investigate the metabolic effects of Dex, C57B6J WT mice were intraperitoneally injected with Dex for 6 weeks; this model has been commonly used to study the role of Dex (1,30). Dex treatment did not change body weight, although the total body fat and abdominal fat mass were increased compared with those in mice receiving the control treatment, possibly as a result of decreased lean mass (Supplementary Fig. 1A–D). A balance between energy intake and energy expenditure maintains body fat mass (7). Dex treatment did not change food intake but did decrease energy expenditure as measured by 24-h indirect calorimetry (Supplementary Fig. 1E and F). No difference was observed in locomotor activity, but body temperature, levels of the BAT thermogenic marker UCP1 (11), and levels of serum NE were significantly lower in Dex-treated mice (Supplementary Fig. 1G–J).

To investigate the possible involvement of hypothalamic SGK1 in Dex-increased adiposity, we examined hypothalamic SGK1 expression under this condition; it is interesting that we found that hypothalamic SGK1 and phosphorylated (p-)SGK1 were decreased in Dex-treated mice (Fig. 1A and B). Furthermore, immunofluorescence staining showed that SGK1 and p-SGK1 were decreased in the ARC of the hypothalamus of Dex-treated mice (Fig. 1C–F). Immunofluorescence staining of tdTomato and SGK1 showed that SGK1 was colocalized with POMC neurons in PBS-treated mice but was decreased significantly in POMC neurons of Dex-treated mice (Fig. 1G and H). By contrast, acute treatment increased SGK1 expression in the ARC of the hypothalamus (Fig. 1I–L).

Figure 1

SGK1 expression in hypothalamic POMC neurons under chronic or acute Dex treatment. A: Sgk1 expression in the hypothalamus under chronic Dex treatment. B: Western blotting (left) and densitometric quantification (right) of SGK1 and p-SGK1 in the hypothalamus. C and D: Immunofluorescence for SGK1 in ARC sections (C) and integrated density quantification (D). E and F: Immunofluorescence for p-SGK1 in ARC sections (E) and integrated density quantification (F). G and H: Immunofluorescence for POMC neurons (red), SGK1 (green), and a merge (yellow) in ARC sections (G) and integrated density quantification in POMC neurons and colocalization (H). I: Sgk1 expression in the hypothalamus under acute Dex treatment. J: Western blotting (left) and densitometric quantification (right) of SGK1 in the hypothalamus under acute Dex treatment. K and L: Immunofluorescence for POMC neurons (red), SGK1 (green), and a merge (yellow) in ARC sections (K) and integrated density quantification in POMC neurons and colocalization (L) under acute Dex treatment. Studies were conducted using 14- to 15-week-old male WT mice (AF), or POMC-tdTomato indicator mice (G and H) treated without Dex (− Dex) or with Dex (+ Dex) every other day for 6 weeks, or in 9-week-old male WT mice (I and J) or POMC-tdTomato indicator mice (for K and L) treated without Dex (− Dex) or with Dex (+ Dex) for 2 h. Data are expressed as the mean ± SEM (n = 6–11 mice/group). *P < 0.05 for the effect of Dex treatment vs. no Dex treatment.

Figure 1

SGK1 expression in hypothalamic POMC neurons under chronic or acute Dex treatment. A: Sgk1 expression in the hypothalamus under chronic Dex treatment. B: Western blotting (left) and densitometric quantification (right) of SGK1 and p-SGK1 in the hypothalamus. C and D: Immunofluorescence for SGK1 in ARC sections (C) and integrated density quantification (D). E and F: Immunofluorescence for p-SGK1 in ARC sections (E) and integrated density quantification (F). G and H: Immunofluorescence for POMC neurons (red), SGK1 (green), and a merge (yellow) in ARC sections (G) and integrated density quantification in POMC neurons and colocalization (H). I: Sgk1 expression in the hypothalamus under acute Dex treatment. J: Western blotting (left) and densitometric quantification (right) of SGK1 in the hypothalamus under acute Dex treatment. K and L: Immunofluorescence for POMC neurons (red), SGK1 (green), and a merge (yellow) in ARC sections (K) and integrated density quantification in POMC neurons and colocalization (L) under acute Dex treatment. Studies were conducted using 14- to 15-week-old male WT mice (AF), or POMC-tdTomato indicator mice (G and H) treated without Dex (− Dex) or with Dex (+ Dex) every other day for 6 weeks, or in 9-week-old male WT mice (I and J) or POMC-tdTomato indicator mice (for K and L) treated without Dex (− Dex) or with Dex (+ Dex) for 2 h. Data are expressed as the mean ± SEM (n = 6–11 mice/group). *P < 0.05 for the effect of Dex treatment vs. no Dex treatment.

Close modal

Deletion of SGK1 in POMC Neurons Causes Obesity and Decreases Energy Expenditure

Based on the above results, we speculated that knockout of SGK1 expression in POMC neurons might mimic Dex-induced metabolic alterations. To test this hypothesis, we generated PSKO mice. Immunofluorescence staining of tdTomato and SGK1 showed that SGK1 was colocalized with POMC neurons (>90% overlap with tdTomato) in control mice but was almost absent in POMC neurons of PSKO mice (Fig. 2A and Supplementary Fig. 2A–C), with no difference in SGK1 staining in the paraventricular nucleus (PVN) and ventromedial hypothalamus (VMH) between PSKO and control mice (Supplementary Fig. 2D–G). Sgk1 mRNA levels were consistently decreased ∼50% in the ARC, as the ARC of PSKO mice have other neurons or neurogliocytes (31,32), but Sgk1 mRNA levels were unchanged in other brain areas and tissues (Fig. 2B). Anatomical assessment of POMC neurons throughout the ARC area revealed no significant alterations in neuronal population size, distribution, or somatic area (Supplementary Fig. 3A and B), indicating that SGK1 deficiency did not alter POMC neuron differentiation and/or survival. Because the POMC promoter also drives CRE recombinase expression in the pituitary (33), we examined serum contents of hormones secreted from the pituitary, including corticosterone and growth hormone (34), and found that the levels of these two hormones were not altered in PSKO mice (Supplementary Fig. 3C and D).

Figure 2

PSKO mice exhibit an obese phenotype and decreased energy expenditure the same as Dex-treated WT mice. A: Immunofluorescence for POMC neurons (red), SGK1 (green), and a merge (yellow) in ARC sections from male POMC-tdTomato indicator mice. B: Sgk1 expression in different tissues. COR, cortex; LV, liver. C: Body weight curve. Graphs show total body fat mass (D), abdominal fat mass (E), daily food intake (F), daily energy expenditure (EE) (G), daily RER (Vco2/Vo2) (H), daily locomotor activity (I), and basal rectal temperature (J). K: Western blotting (top) and densitometric quantification (bottom) of UCP1 in BAT. L: Serum NE. All studies were conducted in 12- to 14-week-old male control (− PSKO) and PSKO (+ PSKO) mice. Data are expressed as the mean ± SEM (n = 6–16 mice/group). *P < 0.05 for the effect of the PSKO group vs. the control group.

Figure 2

PSKO mice exhibit an obese phenotype and decreased energy expenditure the same as Dex-treated WT mice. A: Immunofluorescence for POMC neurons (red), SGK1 (green), and a merge (yellow) in ARC sections from male POMC-tdTomato indicator mice. B: Sgk1 expression in different tissues. COR, cortex; LV, liver. C: Body weight curve. Graphs show total body fat mass (D), abdominal fat mass (E), daily food intake (F), daily energy expenditure (EE) (G), daily RER (Vco2/Vo2) (H), daily locomotor activity (I), and basal rectal temperature (J). K: Western blotting (top) and densitometric quantification (bottom) of UCP1 in BAT. L: Serum NE. All studies were conducted in 12- to 14-week-old male control (− PSKO) and PSKO (+ PSKO) mice. Data are expressed as the mean ± SEM (n = 6–16 mice/group). *P < 0.05 for the effect of the PSKO group vs. the control group.

Close modal

Male PSKO mice exhibited significantly increased body weight from the age of 9 weeks compared with control mice (Fig. 2C); this was accompanied by a significant increase in total body fat and abdominal fat mass (Fig. 2D and E) but unchanged lean mass (Supplementary Fig. 2H). Food intake was not altered, but the energy expenditure was markedly decreased and the respiratory exchange ratio (RER; Vco2/Vo2) was higher in PSKO mice (Fig. 2F–H). Again, locomotor activity was not changed, but body temperature, UCP1 in BAT, and serum NE levels were significantly lower in PSKO mice (Fig. 2I–L). Female PSKO mice displayed phenotypes similar to those observed in male mice (Supplementary Fig. 4), so we performed all of the subsequent studies in male mice.

Inducible Loss of SGK1 in POMC Neurons in Adult Mice Recapitulates Aberrant Energy Homeostasis

We next asked whether adult-onset loss of SGK1 in POMC neurons had effects similar to those of ablation during development. We used the POMC-cre:ERT2 mouse model (24) that allows temporal control of CRE recombinase activity and can be combined with SGK1flox/flox mice to produce mice with adult-onset deletion of SGK1 (PSKO-ER). We observed phenotypes similar to those found when using constitutive POMC-Cre mice (Supplementary Fig. 5).

Mice With SGK1 Overexpression in POMC Neurons Are Lean and Resistant to Dex-Induced Fat Accumulation

We then asked whether overexpression of SGK1 in POMC neurons in mice would have the opposite phenotype of that observed in PSKO mice and whether Dex-increased adiposity would be avoided. For this purpose, we generated PSOE mice by bilateral stereotaxic injection into the ARC of AAV-CA SGK1 or control AAV-null male POMC-Cre mice. The effect of SGK1 overexpression was validated by immunofluorescence staining of the phosphorylated NDRG1, which reflects the activation status of SGK1 (35) (Fig. 3A), and increased SGK1 signals in POMC neurons (>90% overlap with mCherry), but not in the PVN and VMH, of PSOE mice (Supplementary Fig. 6A–H). As predicted, body weight decreased (starting from 6 weeks after AAV injection), accompanied by a decrease in total body fat and abdominal fat mass, in PSOE mice (Fig. 3B–D). Food intake was not affected, but the energy expenditure was increased and RER was decreased in PSOE mice (Fig. 3E–G). No difference was observed in locomotor activity, but body temperature, UCP1 in BAT, and serum NE levels were increased in PSOE mice (Fig. 3H–K). Furthermore, PSOE mice were resistant to Dex-induced fat accumulation and other metabolic alterations (Fig. 4), and Dex injected 5 weeks after AAV injection created no difference in lean mass and fat mass between control and PSOE mice (Supplementary Fig. 6I and J). By contrast, Dex had a very mild effect on PSKO mice, as demonstrated by the slightly decreased body weight and lean mass as well as increased fat mass, and no significant effect on food intake or energy expenditure (Supplementary Fig. 7).

Figure 3

PSOE mice show a lean phenotype and increased energy expenditure. A: Immunofluorescence for POMC neurons (red), p-NDRG1 (green), and a merge (yellow) in ARC sections. Graphs show a body weight curve (B), total body fat mass (C), abdominal fat mass (D), daily food intake (E), daily energy expenditure (EE) (F), daily RER (Vco2/Vo2) (G), daily locomotor activity (H), and basal rectal temperature (I). J: Western blotting (top) and densitometric quantification (bottom) of UCP1 in BAT. K: Serum NE. All studies were conducted in 19- to 20-week-old male control (− PSOE) and PSOE (+ PSOE) mice. Data are expressed as the mean ± SEM (n = 6–9 mice/group). *P < 0.05 for the effect of the PSOE group vs. the control group.

Figure 3

PSOE mice show a lean phenotype and increased energy expenditure. A: Immunofluorescence for POMC neurons (red), p-NDRG1 (green), and a merge (yellow) in ARC sections. Graphs show a body weight curve (B), total body fat mass (C), abdominal fat mass (D), daily food intake (E), daily energy expenditure (EE) (F), daily RER (Vco2/Vo2) (G), daily locomotor activity (H), and basal rectal temperature (I). J: Western blotting (top) and densitometric quantification (bottom) of UCP1 in BAT. K: Serum NE. All studies were conducted in 19- to 20-week-old male control (− PSOE) and PSOE (+ PSOE) mice. Data are expressed as the mean ± SEM (n = 6–9 mice/group). *P < 0.05 for the effect of the PSOE group vs. the control group.

Close modal
Figure 4

PSOE mice are resistant to Dex-induced fat accumulation and decreased energy expenditure. A: Body weight curve. B: Total body fat mass. C: abdominal fat mass. D: Daily food intake. E: Daily energy expenditure (EE). F: Daily RER (Vco2/Vo2). G: Daily locomotor activity. H: Basal rectal temperature. I: Western blotting (left) and densitometric quantification (right) of UCP1 in BAT. J: Serum NE. All studies were conducted in 19- to 20-week-old male control (− PSOE) and PSOE (+ PSOE) mice treated with Dex (+ Dex). Data are expressed as the mean ± SEM (n = 6–12 mice/group). *P < 0.05 for the effect of the PSOE group vs. the control group.

Figure 4

PSOE mice are resistant to Dex-induced fat accumulation and decreased energy expenditure. A: Body weight curve. B: Total body fat mass. C: abdominal fat mass. D: Daily food intake. E: Daily energy expenditure (EE). F: Daily RER (Vco2/Vo2). G: Daily locomotor activity. H: Basal rectal temperature. I: Western blotting (left) and densitometric quantification (right) of UCP1 in BAT. J: Serum NE. All studies were conducted in 19- to 20-week-old male control (− PSOE) and PSOE (+ PSOE) mice treated with Dex (+ Dex). Data are expressed as the mean ± SEM (n = 6–12 mice/group). *P < 0.05 for the effect of the PSOE group vs. the control group.

Close modal

Dex Decreases Hypothalamic α-MSH Content via SGK1, and Administration of α-MSH Reverses Obese Phenotype in PSKO Mice

Because previous studies have shown that α-MSH plays a critical role in the regulation of energy homeostasis (33), we asked whether it might be involved in Dex-induced metabolic alterations. As predicted, a dramatic reduction of α-MSH staining was observed in PVN of Dex-treated mice (Fig. 5A and B). Similar results were obtained in PSKO mice (Fig. 5C and D). The amount of α-MSH, as analyzed by ELISA, was consistently significantly decreased in the hypothalamus of PSKO mice (Fig. 5E). Notably, Dex-reduced α-MSH staining was reversed in PSOE mice (Fig. 5F and G).

Figure 5

Dex decreases hypothalamic α-MSH content via SGK1, and ICV administration of α-MSH reverses the obese phenotype in PSKO mice. A and B: Immunofluorescence for α-MSH in PVN sections (A) of, and integrated density quantification (B) in, 14- to 15-week-old male WT mice treated with no Dex (− Dex) or with Dex (+ Dex). CE: Immunofluorescence for α-MSH in PVN sections (C), integrated density quantification (D), and relative hypothalamic α-MSH content based on ELISA (E) in 12- to 14-week-old male control and PSKO mice. F and G: Immunofluorescence for α-MSH in PVN sections (F) and integrated density quantification (G) in 19- to 20-week-old male control and PSOE mice treated with no Dex (− Dex) or with Dex (+ Dex). HK: Body weight (H), abdominal fat mass (I), basal rectal temperature (J), and Western blotting (top) and densitometric quantification (bottom) of UCP1 in BAT (K) in 10- to 12-week-old male control (− PSKO) and PSKO (+ PSKO) mice treated with no α-MSH (− α-MSH) or with α-MSH (+ α-MSH). Data are expressed as the mean ± SEM (n = 6–8 mice/group). *P < 0.05 for any treatment compared with the control group (AE). *P < 0.05 for the effect of any group vs. control mice treated with no Dex; #P < 0.05 for the effect of PSOE mice vs. control mice after Dex treatment (G). *P < 0.05 for the effect of any group vs. control mice treated with no α-MSH; #P < 0.05 for the effect of treatment with vs. without α-MSH in PSKO mice (HK).

Figure 5

Dex decreases hypothalamic α-MSH content via SGK1, and ICV administration of α-MSH reverses the obese phenotype in PSKO mice. A and B: Immunofluorescence for α-MSH in PVN sections (A) of, and integrated density quantification (B) in, 14- to 15-week-old male WT mice treated with no Dex (− Dex) or with Dex (+ Dex). CE: Immunofluorescence for α-MSH in PVN sections (C), integrated density quantification (D), and relative hypothalamic α-MSH content based on ELISA (E) in 12- to 14-week-old male control and PSKO mice. F and G: Immunofluorescence for α-MSH in PVN sections (F) and integrated density quantification (G) in 19- to 20-week-old male control and PSOE mice treated with no Dex (− Dex) or with Dex (+ Dex). HK: Body weight (H), abdominal fat mass (I), basal rectal temperature (J), and Western blotting (top) and densitometric quantification (bottom) of UCP1 in BAT (K) in 10- to 12-week-old male control (− PSKO) and PSKO (+ PSKO) mice treated with no α-MSH (− α-MSH) or with α-MSH (+ α-MSH). Data are expressed as the mean ± SEM (n = 6–8 mice/group). *P < 0.05 for any treatment compared with the control group (AE). *P < 0.05 for the effect of any group vs. control mice treated with no Dex; #P < 0.05 for the effect of PSOE mice vs. control mice after Dex treatment (G). *P < 0.05 for the effect of any group vs. control mice treated with no α-MSH; #P < 0.05 for the effect of treatment with vs. without α-MSH in PSKO mice (HK).

Close modal

To investigate whether α-MSH could mediate SGK1 regulation of energy homeostasis, we administered α-MSH peptide ICV to PSKO or control mice. ICV injection of α-MSH to PSKO mice markedly reduced body weight and abdominal fat mass and increased rectal temperature compared with those values in mice injected with the vehicle (control; Fig. 5H–J). ICV injection of α-MSH in PSKO mice also blocked a UCP1 protein decrease (Fig. 5K). Similar effects were observed in control mice after ICV injection of α-MSH (Fig. 5H–K).

Dex Reduces α-MSH Precursor POMC Expression Via the SGK1/FOXO3-Dependent Pathway, and Downregulation of FOXO3 Largely Reverses the Obesity Phenotype in PSKO Mice

α-MSH levels are determined by the levels of its precursor POMC and the expression of prohormone convertases that are responsible for cleaving POMC to α-MSH (8). The reduced α-MSH concentration in Dex-treated mice did not seem to be a consequence of decreased expression of processing enzymes, including prohormone convertase 1 (Pc1/3), prohormone convertase 2 (Pc2), carboxypeptidase E (Cpe), α-amidating monooxygenase (Pam), and prolylcarboxypeptidase (Prcp) (8), as gene expression of these enzymes was unchanged (Fig. 6A). On the other hand, POMC expression was decreased in Dex-treated mice (Fig. 6A–C). Similar results were obtained in PSKO mice (Supplementary Fig. 8A–C). The effect of Dex on reducing POMC expression, however, was reversed by overexpression of SGK1 (Fig. 6D and E). Similarly, SGK1 knockdown decreased Pomc expression and SGK1 overexpression increased Pomc expression in primary cultured hypothalamic neurons (Supplementary Fig. 8D and E).

Figure 6

Dex reduces α-MSH precursor POMC expression via the SGK1/FOXO3-dependent pathway, and downregulation of FOXO3 in the ARC largely reverses the obesity phenotype in PSKO mice. AC: Neuropeptide expression in the hypothalamus (A), immunofluorescence for POMC in ARC sections (B), and integrated density quantification (C) in 14- to 15-week-old male WT mice treated with no Dex (− Dex) or with Dex (+ Dex). D and E: Immunofluorescence for POMC in ARC sections (D) and integrated density quantification (E) in 19- to 20-week-old male control and PSOE mice treated with no Dex (− Dex) or with Dex (+ Dex). F: Western blotting (top) and densitometric quantification (bottom) of p-FOXO3 and FOXO3 in the hypothalamus in 14- to 15-week-old male WT mice treated with no Dex (– Dex) or with Dex (+ Dex). G and H: Immunofluorescence for POMC neurons (red), p-FOXO3 (green), and a merge (yellow) in ARC sections (G) and integrated density quantification in POMC neurons and colocalization (H) in 19- to 20-week-old male control and PSOE mice treated with no Dex (− Dex) or with Dex (+ Dex). IQ: The expression of Sgk1, Foxo3, and Pomc in the ARC (I), body weight (J), total body fat mass (K), abdominal fat mass (L), daily energy expenditure (EE) (M), daily RER (Vco2/Vo2) (N), basal rectal temperature (O), Western blotting (top) and densitometric quantification (bottom) of UCP1 in BAT (P), and serum NE (Q) in 16- to 18-week-old male control (− PSKO) and PSKO (+ PSKO) mice injected with Ad-scrambled (− Ad-shFOXO3) or Ad-shFOXO3 (+ Ad-shFOXO3). Data are expressed as the mean ± SEM (n = 6–11 mice/group). *P < 0.05 for the effect of Dex treatment vs. no Dex treatment (A, C, and F). *P < 0.05 for the effect of any group vs. control mice treated with no Dex; #P < 0.05 for the effect of PSOE mice vs. control mice after Dex treatment (E and H). *P < 0.05 for the effect of any group vs. control mice not injected with Ad-shFOXO3; #P < 0.05 for the effect of Ad-shFOXO3 injection vs. no Ad-shFOXO3 injection in PSKO mice (IQ).

Figure 6

Dex reduces α-MSH precursor POMC expression via the SGK1/FOXO3-dependent pathway, and downregulation of FOXO3 in the ARC largely reverses the obesity phenotype in PSKO mice. AC: Neuropeptide expression in the hypothalamus (A), immunofluorescence for POMC in ARC sections (B), and integrated density quantification (C) in 14- to 15-week-old male WT mice treated with no Dex (− Dex) or with Dex (+ Dex). D and E: Immunofluorescence for POMC in ARC sections (D) and integrated density quantification (E) in 19- to 20-week-old male control and PSOE mice treated with no Dex (− Dex) or with Dex (+ Dex). F: Western blotting (top) and densitometric quantification (bottom) of p-FOXO3 and FOXO3 in the hypothalamus in 14- to 15-week-old male WT mice treated with no Dex (– Dex) or with Dex (+ Dex). G and H: Immunofluorescence for POMC neurons (red), p-FOXO3 (green), and a merge (yellow) in ARC sections (G) and integrated density quantification in POMC neurons and colocalization (H) in 19- to 20-week-old male control and PSOE mice treated with no Dex (− Dex) or with Dex (+ Dex). IQ: The expression of Sgk1, Foxo3, and Pomc in the ARC (I), body weight (J), total body fat mass (K), abdominal fat mass (L), daily energy expenditure (EE) (M), daily RER (Vco2/Vo2) (N), basal rectal temperature (O), Western blotting (top) and densitometric quantification (bottom) of UCP1 in BAT (P), and serum NE (Q) in 16- to 18-week-old male control (− PSKO) and PSKO (+ PSKO) mice injected with Ad-scrambled (− Ad-shFOXO3) or Ad-shFOXO3 (+ Ad-shFOXO3). Data are expressed as the mean ± SEM (n = 6–11 mice/group). *P < 0.05 for the effect of Dex treatment vs. no Dex treatment (A, C, and F). *P < 0.05 for the effect of any group vs. control mice treated with no Dex; #P < 0.05 for the effect of PSOE mice vs. control mice after Dex treatment (E and H). *P < 0.05 for the effect of any group vs. control mice not injected with Ad-shFOXO3; #P < 0.05 for the effect of Ad-shFOXO3 injection vs. no Ad-shFOXO3 injection in PSKO mice (IQ).

Close modal

We then investigated the downstream signaling of SGK1 in mediating Dex-decreased POMC expression. Previous studies showed that SGK1 phosphorylates FOXO3 (36) and that another member from the same FOXO family, FOXO1, inhibits Pomc expression (37), suggesting that FOXO3 might have a function similar to that of FOXO1 downstream of SGK1 in Dex-induced metabolic alterations. Consistent with this possibility, hypothalamic FOXO3 phosphorylation was decreased in Dex-treated mice (Fig. 6F). Similar reduction was observed in the hypothalamic ARC of PSKO mice (Supplementary Fig. 9A). Furthermore, Dex-decreased hypothalamic FOXO3 phosphorylation was reversed in PSOE mice (Fig. 6G and H). Similar regulatory effects of SGK1 on p-FOXO3 were observed in primary cultures of hypothalamic neurons (Supplementary Fig. 9B and C).

The inhibitory effect of SGK1 knockdown on Pomc expression was reversed by small interfering RNA–mediated FOXO3 inhibition (Supplementary Fig. 9D), thereby prompting us to investigate the in vivo function of FOXO3 downstream of SGK1. For this purpose, we knocked down FOXO3 expression in the ARC of PSKO and control mice by ARC administration (6) of adenovirus expressing short hairpin RNA directed against the coding region of FOXO3 (Ad-shFOXO3) (38) or Ad-scrambled. The effect of Ad-shFOXO3 was demonstrated through decreased expression of Foxo3 and the corresponding change in Pomc expression in the ARC of PSKO mice (Fig. 6I). Immunofluorescence consistently showed that FOXO3 was decreased in the ARC, but not the PVN or VMH, of these mice (Supplementary Fig. 10A and B). Ad-shFOXO3 decreased the body weight, total body fat, and abdominal fat mass in PSKO mice (Fig. 6J–L). Although food intake was not affected (Supplementary Fig. 10C), the decreased energy expenditure and increased RER in PSKO mice were largely reversed by Ad-shFOXO3 (Fig. 6M and N). No significant difference in locomotor activity was detected (Supplementary Fig. 10D); however, the decreased body temperature, UCP1 in BAT, and serum NE in PSKO mice were upregulated by Ad-shFOXO3 (Fig. 6O–Q). Moreover, the reduced α-MSH staining in PSKO mice was also blocked by Ad-shFOXO3 (Supplementary Fig. 10E and F). Except for the unaltered body weight, similar effects were observed in control mice following administration of Ad-shFOXO3 (Fig. 6I–Q and Supplementary Fig. 10).

Because glucocorticoid functions via the GR (28), we investigated the spatial regulation of GR and SGK1/FOXO3 using previously validated GR antibodies (39). Although hypothalamic Gr mRNA was unchanged, total GR and phosphorylated GR expression were significantly decreased in Dex-treated mice (Supplementary Fig. 11A and B). Furthermore, these three proteins were all expressed in POMC neurons and hypothalamic nuclear p-GR was decreased and FOXO3 was increased, but the cytoplasmic total and phosphorylated proteins examined were all decreased, in Dex-treated mice (Supplementary Fig. 11C and D).

Fat mass accumulation is a serious side effect of glucocorticoid therapy (1). Recent studies have elucidated several peripheral mechanisms underlying glucocorticoid-induced gains in fat mass (15). In this study we demonstrated a novel central mechanism, mediated by SGK1, underlying glucocorticoid-increased adiposity. SGK1 is a well-known downstream target of Dex (2022). It has been widely demonstrated that acute Dex treatment induces SGK1 (2022). It is interesting to note that we found decreased SGK1 expression in POMC neurons in the ARC of Dex-treated mice. The importance of POMC SGK1 in mediating Dex-induced adiposity was demonstrated by the observation that knockout of SGK1 expression in POMC neurons increased adiposity, whereas overexpression of SGK1 in POMC neurons resulted in a lean phenotype and prevented Dex-induced fat mass gain in mice. Furthermore, the Dex-induced gain in fat mass was much lower in PSKO than in control mice. Fat mass could, however, still be increased by Dex treatment in PSKO mice, suggesting the existence of other central or peripheral signals involved in Dex-increased adiposity (40). Our study provides a novel mechanistic link between glucocorticoid treatment and fat mass gain. This is important for understanding the mechanisms of glucocorticoid-induced metabolic phenotypes and provides an important hint for a possible treatment target for glucocorticoid-induced side effects. In addition, our study reports an unrecognized novel function of SGK1 in POMC neurons of the hypothalamus in the regulation of energy homeostasis. These results are important for understanding the signals in specific neurons that are critical for metabolic control.

A balance between energy intake and energy expenditure maintains body fat mass (7). BAT oxidizes fat to produce heat via increased expression of uncoupled proteins, which is stimulated by the activation of the SNS. Deletion of UCP1 induces obesity and upregulation of UCP1 increases thermogenesis and energy expenditure in mice (11). Other studies also showed that disruption of SNS activity has a significant negative impact on energy expenditure (6,41,42). Our study showed that Dex increased adiposity mainly by decreasing energy expenditure, as food intake was not changed in Dex-treated mice. Furthermore, the decrease in energy expenditure resulting from Dex treatment was most likely due to decreased thermogenesis in BAT, as demonstrated by the decreased body temperature, UCP1 expression in BAT, and serum NE in these mice. Lipolysis in WAT is also regulated by SNS activity (41,42), which might also affect Dex-induced adiposity and should be studied in the future.

Extensive evidence indicates that melanocortin signaling in the hypothalamus plays an important role in regulating energy homeostasis and lipid metabolism through its effects on SNS activity in BAT (1114). In this study we demonstrated a possible role for α-MSH in mediating Dex regulation of adiposity, as α-MSH levels were decreased in Dex-treated mice via SGK1, and restoration of hypothalamic α-MSH levels by ICV administration of this peptide normalized inadequate energy homeostasis in PSKO mice. Although the beneficial effects of this pharmacological treatment are most likely mediated through direct actions on POMC neurons, we cannot exclude its potential effects on other hypothalamic areas as a result of the delivery route used.

Our results suggest that the reduced α-MSH content in Dex-treated mice was not caused by an altered proteolysis process, but the decreased Pomc expression is possibly due to glucocorticoid resistance, as Dex was shown to induce Pomc expression (43). Furthermore, we found that Dex decreased Pomc expression via the SGK1/FOXO3-dependent pathway, as the inhibitory effect of Dex on Pomc expression was blocked in mice with SGK1 overexpression or FOXO3 inhibition. Many studies, including those conducted on FOXO3 knockout mice, have demonstrated that FOXO3 is vital for many functions of the CNS and has roles in neural stem cell homeostasis, stress, and Huntington disease (44,45). We showed that it functions as a downstream signal of SGK1 in the regulation of energy homeostasis. We also demonstrated the spatial relationships among GR, SGK1, and FOXO3, providing a basis for the interaction among and regulation of these proteins.

In this study we also demonstrated that adult-onset loss of SGK1 in POMC neurons results in a phenotype similar to that of ablation during development. This is a key issue, because some works report that multiple hypothalamic neurons express POMC in adult mice (24) and prenatal and postnatal ablation of certain neurons result in disparate feeding behavior, suggesting that phenotypes caused by prenatal ablation may be influenced by developmental compensation (24). The POMC promoter also drives CRE recombinase expression in corticotrophs and melanotrophs (46). The contribution of the pituitary might not be that significant in this study, as no changes were observed in serum corticosterone and growth hormone, which reflect the function of the pituitary (34), between PSKO and control mice.

Previous studies have shown that POMC neurons are involved in the regulation of food intake (29,47). For reasons unknown, however, we found that food intake was not significantly affected by Dex treatment, or in PSKO or PSOE mice. Consistent with our study, however, previous works indicate that genetic blockade of the CNS–melanocortin 3 receptor promotes fat accumulation in the absence of hyperphagia (48).

In contrast to the stimulatory effect of glucocorticoid on SGK1 expression (2022), we observed decreased hypothalamic SGK1 expression following chronic Dex treatment, which is, to our knowledge, a novel observation. We speculate that this inhibition is not a direct effect of Dex on SGK1 expression but rather a consequence of attenuated Dex-mediated signaling, as it has been previously shown that prolonged Dex treatment causes glucocorticoid resistance (49). Because glucocorticoid normally functions via the GR (28), the difference in SGK1 expression under acute or chronic Dex treatment may be caused by differences in GR activity under different conditions, as shown by our work and that of others (28,39). In addition, because chronic Dex treatment affects the activity of several regulatory molecules that influence SGK1 transcription and/or mRNA decay (28,50), and because hypothalamic signals might also be affected by peripheral events (6,16,26), the possible contribution of these factors to hypothalamic SGK1 expression cannot be excluded in Dex-treated mice. These possibilities will be explored in future studies.

In summary, our results demonstrate that SGK1/FOXO3 signaling in POMC neurons is crucial for Dex-induced adiposity. These results provide novel insights into the central mechanisms underlying Dex-induced obesity. In this study we also established that SGK1 in POMC neurons is an essential regulator of systemic energy balance. This previously unrecognized role for hypothalamic SGK1 also indicates a potential novel drug target in treating obesity and its related metabolic disorders.

Funding. This work was supported by grants from the National Natural Science Foundation of China (81325005, 81390350, 81471076, 81570777, 81130076, 31271269, 81400792, 81500622, and 81600623), a Basic Research Project of the Shanghai Science and Technology Commission (16JC1404900 and 17XD1404200), and the Chinese Academy of Sciences/State Administration of Foreign Experts Affairs International Partnership Program for Creative Research Teams. F.G. was supported by the One Hundred Talents Program of the Chinese Academy of Sciences.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. Y.D. researched data and wrote, reviewed, and edited the manuscript. Y.X., F.Y., Y.L., X.J., and J.D. researched data. G.F.-T. and A.N.-F.-T. generated and provided the floxed SGK1 mice. S.C. provided research material. Y.C., H.Y., and Q.Z. directed the project and contributed to the discussion. Y.S. and F.G. directed the project; contributed to the discussion; and wrote, reviewed, and edited the manuscript. Y.S. and F.G. are the guarantors of this work and, as such, had full access to all the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Ma
X
,
Xu
L
,
Mueller
E
.
Forkhead box A3 mediates glucocorticoid receptor function in adipose tissue
.
Proc Natl Acad Sci U S A
2016
;
113
:
3377
3382
[PubMed]
2.
Cha
JY
,
Kim
HJ
,
Yu
JH
, et al
.
Dexras1 mediates glucocorticoid-associated adipogenesis and diet-induced obesity
.
Proc Natl Acad Sci U S A
2013
;
110
:
20575
20580
[PubMed]
3.
Chapman
AB
,
Knight
DM
,
Ringold
GM
.
Glucocorticoid regulation of adipocyte differentiation: hormonal triggering of the developmental program and induction of a differentiation-dependent gene
.
J Cell Biol
1985
;
101
:
1227
1235
[PubMed]
4.
Kong
X
,
Yu
J
,
Bi
J
, et al
.
Glucocorticoids transcriptionally regulate miR-27b expression promoting body fat accumulation via suppressing the browning of white adipose tissue
.
Diabetes
2015
;
64
:
393
404
[PubMed]
5.
Soumano
K
,
Desbiens
S
,
Rabelo
R
,
Bakopanos
E
,
Camirand
A
,
Silva
JE
.
Glucocorticoids inhibit the transcriptional response of the uncoupling protein-1 gene to adrenergic stimulation in a brown adipose cell line
.
Mol Cell Endocrinol
2000
;
165
:
7
15
[PubMed]
6.
Schneeberger
M
,
Dietrich
MO
,
Sebastián
D
, et al
.
Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalance
.
Cell
2013
;
155
:
172
187
[PubMed]
7.
Schwartz
MW
,
Porte
D
 Jr
.
Diabetes, obesity, and the brain
.
Science
2005
;
307
:
375
379
[PubMed]
8.
Wardlaw
SL
.
Hypothalamic proopiomelanocortin processing and the regulation of energy balance
.
Eur J Pharmacol
2011
;
660
:
213
219
[PubMed]
9.
Huszar
D
,
Lynch
CA
,
Fairchild-Huntress
V
, et al
.
Targeted disruption of the melanocortin-4 receptor results in obesity in mice
.
Cell
1997
;
88
:
131
141
[PubMed]
10.
Farooqi
IS
,
Keogh
JM
,
Yeo
GS
,
Lank
EJ
,
Cheetham
T
,
O’Rahilly
S
.
Clinical spectrum of obesity and mutations in the melanocortin 4 receptor gene
.
N Engl J Med
2003
;
348
:
1085
1095
[PubMed]
11.
Lowell
BB
,
Spiegelman
BM
.
Towards a molecular understanding of adaptive thermogenesis
.
Nature
2000
;
404
:
652
660
[PubMed]
12.
Holm
C
.
Molecular mechanisms regulating hormone-sensitive lipase and lipolysis
.
Biochem Soc Trans
2003
;
31
:
1120
1124
[PubMed]
13.
Hücking
K
,
Hamilton-Wessler
M
,
Ellmerer
M
,
Bergman
RN
.
Burst-like control of lipolysis by the sympathetic nervous system in vivo
.
J Clin Invest
2003
;
111
:
257
264
[PubMed]
14.
Morrison
SF
,
Madden
CJ
,
Tupone
D
.
Central neural regulation of brown adipose tissue thermogenesis and energy expenditure
.
Cell Metab
2014
;
19
:
741
756
[PubMed]
15.
Veyrat-Durebex
C
,
Deblon
N
,
Caillon
A
, et al
.
Central glucocorticoid administration promotes weight gain and increased 11β-hydroxysteroid dehydrogenase type 1 expression in white adipose tissue
.
PLoS One
2012
;
7
:
e34002
[PubMed]
16.
Zakrzewska
KE
,
Cusin
I
,
Stricker-Krongrad
A
, et al
.
Induction of obesity and hyperleptinemia by central glucocorticoid infusion in the rat
.
Diabetes
1999
;
48
:
365
370
[PubMed]
17.
Lang
F
,
Artunc
F
,
Vallon
V
.
The physiological impact of the serum and glucocorticoid-inducible kinase SGK1
.
Curr Opin Nephrol Hypertens
2009
;
18
:
439
448
[PubMed]
18.
Fejes-Tóth
G
,
Frindt
G
,
Náray-Fejes-Tóth
A
,
Palmer
LG
.
Epithelial Na+ channel activation and processing in mice lacking SGK1
.
Am J Physiol Renal Physiol
2008
;
294
:
F1298
F1305
[PubMed]
19.
Liu
H
,
Yu
J
,
Xia
T
, et al
.
Hepatic serum- and glucocorticoid-regulated protein kinase 1 (SGK1) regulates insulin sensitivity in mice via extracellular-signal-regulated kinase 1/2 (ERK1/2)
.
Biochem J
2014
;
464
:
281
289
[PubMed]
20.
Anacker
C
,
Cattaneo
A
,
Musaelyan
K
, et al
.
Role for the kinase SGK1 in stress, depression, and glucocorticoid effects on hippocampal neurogenesis
.
Proc Natl Acad Sci U S A
2013
;
110
:
8708
8713
[PubMed]
21.
Ullrich
S
,
Berchtold
S
,
Ranta
F
, et al
.
Serum- and glucocorticoid-inducible kinase 1 (SGK1) mediates glucocorticoid-induced inhibition of insulin secretion
.
Diabetes
2005
;
54
:
1090
1099
[PubMed]
22.
van Gemert
NG
,
Meijer
OC
,
Morsink
MC
,
Joëls
M
.
Effect of brief corticosterone administration on SGK1 and RGS4 mRNA expression in rat hippocampus
.
Stress
2006
;
9
:
165
170
[PubMed]
23.
Reiter
MH
,
Vila
G
,
Knosp
E
, et al
.
Opposite effects of serum- and glucocorticoid-regulated kinase-1 and glucocorticoids on POMC transcription and ACTH release
.
Am J Physiol Endocrinol Metab
2011
;
301
:
E336
E341
[PubMed]
24.
Berglund
ED
,
Liu
C
,
Sohn
JW
, et al
.
Serotonin 2C receptors in pro-opiomelanocortin neurons regulate energy and glucose homeostasis [published correction appears in J Clin Invest 2014;124:1868]
.
J Clin Invest
2013
;
123
:
5061
5070
[PubMed]
25.
Hoekstra
M
,
Meurs
I
,
Koenders
M
, et al
.
Absence of HDL cholesteryl ester uptake in mice via SR-BI impairs an adequate adrenal glucocorticoid-mediated stress response to fasting
.
J Lipid Res
2008
;
49
:
738
745
[PubMed]
26.
Kleinridders
A
,
Schenten
D
,
Könner
AC
, et al
.
MyD88 signaling in the CNS is required for development of fatty acid-induced leptin resistance and diet-induced obesity
.
Cell Metab
2009
;
10
:
249
259
[PubMed]
27.
Xia
T
,
Cheng
Y
,
Zhang
Q
, et al
.
S6K1 in the central nervous system regulates energy expenditure via MC4R/CRH pathways in response to deprivation of an essential amino acid
.
Diabetes
2012
;
61
:
2461
2471
[PubMed]
28.
Arango-Lievano
M
,
Lambert
WM
,
Bath
KG
,
Garabedian
MJ
,
Chao
MV
,
Jeanneteau
F
.
Neurotrophic-priming of glucocorticoid receptor signaling is essential for neuronal plasticity to stress and antidepressant treatment
.
Proc Natl Acad Sci U S A
2015
;
112
:
15737
15742
[PubMed]
29.
Koch
M
,
Varela
L
,
Kim
JG
, et al
.
Hypothalamic POMC neurons promote cannabinoid-induced feeding
.
Nature
2015
;
519
:
45
50
[PubMed]
30.
Poggioli
R
,
Ueta
CB
,
Drigo
RA
,
Castillo
M
,
Fonseca
TL
,
Bianco
AC
.
Dexamethasone reduces energy expenditure and increases susceptibility to diet-induced obesity in mice
.
Obesity (Silver Spring)
2013
;
21
:
E415
E420
[PubMed]
31.
Morton
GJ
,
Cummings
DE
,
Baskin
DG
,
Barsh
GS
,
Schwartz
MW
.
Central nervous system control of food intake and body weight
.
Nature
2006
;
443
:
289
295
[PubMed]
32.
Zhang
G
,
Li
J
,
Purkayastha
S
, et al
.
Hypothalamic programming of systemic ageing involving IKK-β, NF-κB and GnRH
.
Nature
2013
;
497
:
211
216
[PubMed]
33.
Plum
L
,
Lin
HV
,
Dutia
R
, et al
.
The obesity susceptibility gene Cpe links FoxO1 signaling in hypothalamic pro-opiomelanocortin neurons with regulation of food intake
.
Nat Med
2009
;
15
:
1195
1201
[PubMed]
34.
Wilson
CA
,
Buckingham
JC
,
Morris
ID
.
Influence of growth hormone, corticosterone, corticotrophin and changes in the environmental temperature on pituitary-ovarian function in the immature rat
.
J Endocrinol
1985
;
104
:
179
183
[PubMed]
35.
Murray
JT
,
Campbell
DG
,
Morrice
N
, et al
.
Exploitation of KESTREL to identify NDRG family members as physiological substrates for SGK1 and GSK3
.
Biochem J
2004
;
384
:
477
488
[PubMed]
36.
Brunet
A
,
Park
J
,
Tran
H
,
Hu
LS
,
Hemmings
BA
,
Greenberg
ME
.
Protein kinase SGK mediates survival signals by phosphorylating the forkhead transcription factor FKHRL1 (FOXO3a)
.
Mol Cell Biol
2001
;
21
:
952
965
[PubMed]
37.
Ma
W
,
Fuentes
G
,
Shi
X
,
Verma
C
,
Radda
GK
,
Han
W
.
FoxO1 negatively regulates leptin-induced POMC transcription through its direct interaction with STAT3
.
Biochem J
2015
;
466
:
291
298
[PubMed]
38.
Mammucari
C
,
Milan
G
,
Romanello
V
, et al
.
FoxO3 controls autophagy in skeletal muscle in vivo
.
Cell Metab
2007
;
6
:
458
471
[PubMed]
39.
Sarabdjitsingh
RA
,
Meijer
OC
,
de Kloet
ER
.
Specificity of glucocorticoid receptor primary antibodies for analysis of receptor localization patterns in cultured cells and rat hippocampus
.
Brain Res
2010
;
1331
:
1
11
[PubMed]
40.
Smart
JL
,
Tolle
V
,
Low
MJ
.
Glucocorticoids exacerbate obesity and insulin resistance in neuron-specific proopiomelanocortin-deficient mice
.
J Clin Invest
2006
;
116
:
495
505
[PubMed]
41.
Kaushik
S
,
Arias
E
,
Kwon
H
, et al
.
Loss of autophagy in hypothalamic POMC neurons impairs lipolysis
.
EMBO Rep
2012
;
13
:
258
265
[PubMed]
42.
Nogueiras
R
,
Wiedmer
P
,
Perez-Tilve
D
, et al
.
The central melanocortin system directly controls peripheral lipid metabolism
.
J Clin Invest
2007
;
117
:
3475
3488
[PubMed]
43.
Wardlaw
SL
,
McCarthy
KC
,
Conwell
IM
.
Glucocorticoid regulation of hypothalamic proopiomelanocortin
.
Neuroendocrinology
1998
;
67
:
51
57
[PubMed]
44.
Scarpa
JR
,
Jiang
P
,
Losic
B
, et al
.
Systems genetic analyses highlight a TGFβ-FOXO3 dependent striatal astrocyte network conserved across species and associated with stress, sleep, and Huntington’s disease
.
PLoS Genet
2016
;
12
:
e1006137
[PubMed]
45.
Renault
VM
,
Rafalski
VA
,
Morgan
AA
, et al
.
FoxO3 regulates neural stem cell homeostasis
.
Cell Stem Cell
2009
;
5
:
527
539
[PubMed]
46.
Stefaneanu
L
,
Kovacs
K
,
Horvath
E
,
Lloyd
RV
.
In situ hybridization study of pro-opiomelanocortin (POMC) gene expression in human pituitary corticotrophs and their adenomas
.
Virchows Arch A Pathol Anat Histopathol
1991
;
419
:
107
113
[PubMed]
47.
Yang
Y
,
Atasoy
D
,
Su
HH
,
Sternson
SM
.
Hunger states switch a flip-flop memory circuit via a synaptic AMPK-dependent positive feedback loop
.
Cell
2011
;
146
:
992
1003
[PubMed]
48.
Chen
AS
,
Marsh
DJ
,
Trumbauer
ME
, et al
.
Inactivation of the mouse melanocortin-3 receptor results in increased fat mass and reduced lean body mass
.
Nat Genet
2000
;
26
:
97
102
[PubMed]
49.
Sousa e Silva
T
,
Longui
CA
,
Rocha
MN
, et al
.
Prolonged physical training decreases mRNA levels of glucocorticoid receptor and inflammatory genes
.
Horm Res Paediatr
2010
;
74
:
6
14
[PubMed]
50.
Cho
H
,
Park
OH
,
Park
J
, et al
.
Glucocorticoid receptor interacts with PNRC2 in a ligand-dependent manner to recruit UPF1 for rapid mRNA degradation
.
Proc Natl Acad Sci U S A
2015
;
112
:
E1540
E1549
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.

Supplementary data